Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nature ; 612(7940): 540-545, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36323336

RESUMEN

The BA.2 sublineage of the SARS-CoV-2 Omicron variant has become dominant in most countries around the world; however, the prevalence of BA.4 and BA.5 is increasing rapidly in several regions. BA.2 is less pathogenic in animal models than previously circulating variants of concern1-4. Compared with BA.2, however, BA.4 and BA.5 possess additional substitutions in the spike protein, which play a key role in viral entry, raising concerns that the replication capacity and pathogenicity of BA.4 and BA.5 are higher than those of BA.2. Here we have evaluated the replicative ability and pathogenicity of BA.4 and BA.5 isolates in wild-type Syrian hamsters, human ACE2 (hACE2) transgenic hamsters and hACE2 transgenic mice. We have observed no obvious differences among BA.2, BA.4 and BA.5 isolates in growth ability or pathogenicity in rodent models, and less pathogenicity compared to a previously circulating Delta (B.1.617.2 lineage) isolate. In addition, in vivo competition experiments revealed that BA.5 outcompeted BA.2 in hamsters, whereas BA.4 and BA.2 exhibited similar fitness. These findings suggest that BA.4 and BA.5 clinical isolates have similar pathogenicity to BA.2 in rodents and that BA.5 possesses viral fitness superior to that of BA.2.


Asunto(s)
COVID-19 , Aptitud Genética , Roedores , SARS-CoV-2 , Animales , Cricetinae , Humanos , Ratones , COVID-19/virología , Mesocricetus/virología , Ratones Transgénicos , Roedores/virología , SARS-CoV-2/clasificación , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Animales Modificados Genéticamente , Aptitud Genética/genética , Aptitud Genética/fisiología , Virulencia
2.
Nature ; 607(7917): 119-127, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35576972

RESUMEN

The recent emergence of SARS-CoV-2 Omicron (B.1.1.529 lineage) variants possessing numerous mutations has raised concerns of decreased effectiveness of current vaccines, therapeutic monoclonal antibodies and antiviral drugs for COVID-19 against these variants1,2. The original Omicron lineage, BA.1, prevailed in many countries, but more recently, BA.2 has become dominant in at least 68 countries3. Here we evaluated the replicative ability and pathogenicity of authentic infectious BA.2 isolates in immunocompetent and human ACE2-expressing mice and hamsters. In contrast to recent data with chimeric, recombinant SARS-CoV-2 strains expressing the spike proteins of BA.1 and BA.2 on an ancestral WK-521 backbone4, we observed similar infectivity and pathogenicity in mice and hamsters for BA.2 and BA.1, and less pathogenicity compared with early SARS-CoV-2 strains. We also observed a marked and significant reduction in the neutralizing activity of plasma from individuals who had recovered from COVID-19 and vaccine recipients against BA.2 compared to ancestral and Delta variant strains. In addition, we found that some therapeutic monoclonal antibodies (REGN10987 plus REGN10933, COV2-2196 plus COV2-2130, and S309) and antiviral drugs (molnupiravir, nirmatrelvir and S-217622) can restrict viral infection in the respiratory organs of BA.2-infected hamsters. These findings suggest that the replication and pathogenicity of BA.2 is similar to that of BA.1 in rodents and that several therapeutic monoclonal antibodies and antiviral compounds are effective against Omicron BA.2 variants.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/farmacología , Anticuerpos Antivirales/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/genética , COVID-19/inmunología , COVID-19/virología , Cricetinae , Citidina/análogos & derivados , Combinación de Medicamentos , Hidroxilaminas , Indazoles , Lactamas , Leucina , Ratones , Nitrilos , Prolina , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/genética , Triazinas , Triazoles
3.
Nature ; 603(7902): 687-692, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35062015

RESUMEN

The recent emergence of B.1.1.529, the Omicron variant1,2, has raised concerns of escape from protection by vaccines and therapeutic antibodies. A key test for potential countermeasures against B.1.1.529 is their activity in preclinical rodent models of respiratory tract disease. Here, using the collaborative network of the SARS-CoV-2 Assessment of Viral Evolution (SAVE) programme of the National Institute of Allergy and Infectious Diseases (NIAID), we evaluated the ability of several B.1.1.529 isolates to cause infection and disease in immunocompetent and human ACE2 (hACE2)-expressing mice and hamsters. Despite modelling data indicating that B.1.1.529 spike can bind more avidly to mouse ACE2 (refs. 3,4), we observed less infection by B.1.1.529 in 129, C57BL/6, BALB/c and K18-hACE2 transgenic mice than by previous SARS-CoV-2 variants, with limited weight loss and lower viral burden in the upper and lower respiratory tracts. In wild-type and hACE2 transgenic hamsters, lung infection, clinical disease and pathology with B.1.1.529 were also milder than with historical isolates or other SARS-CoV-2 variants of concern. Overall, experiments from the SAVE/NIAID network with several B.1.1.529 isolates demonstrate attenuated lung disease in rodents, which parallels preliminary human clinical data.


Asunto(s)
COVID-19/patología , COVID-19/virología , Modelos Animales de Enfermedad , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Cricetinae , Femenino , Humanos , Pulmón/patología , Pulmón/virología , Masculino , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Carga Viral
4.
Microbiol Immunol ; 68(4): 130-147, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38294180

RESUMEN

Vaccination is an important factor in public health. The recombinant bacillus Calmette Guérin (rBCG) vaccine, which expresses foreign antigens, is expected to be a superior vaccine against infectious diseases. Here, we report a new recombination platform in which the BCG Tokyo strain is transformed with nucleotide sequences encoding foreign protein fused with the MPB70 immunogenic protein precursor. By RNA-sequencing, mpb70 was found to be the most transcribed among all known genes of BCG Tokyo. Small oligopeptide, namely, polyhistidine tag, was able to be expressed in and secreted from rBCG through a process in which polyhistidine tag fused with intact MPB70 were transcribed by an mpb70 promoter. This methodology was applied to develop an rBCG expressing the receptor binding domain (RBD) of severe acute respiratory syndrome coronavirus 2. Immunoblotting images and mass spectrometry data showed that RBD was also secreted from rBCG. Sera from mice vaccinated with the rBCG showed a tendency of weak neutralizing capacity. The secretion was retained even after a freeze-drying process. The freeze-dried rBCG was administered to and recovered from mice. Recovered rBCG kept secreting RBD. Collectively, our recombination platform offers stable secretion of foreign antigens and can be applied to the development of practical rBCGs.


Asunto(s)
Vacuna BCG , Mycobacterium bovis , Animales , Ratones , Vacuna BCG/genética , Tokio , Mycobacterium bovis/genética , Activación de Linfocitos , Ingeniería Genética , Vacunas Sintéticas
5.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34140350

RESUMEN

The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) plays a key role in viral infectivity. It is also the major antigen stimulating the host's protective immune response, specifically, the production of neutralizing antibodies. Recently, a new variant of SARS-CoV-2 possessing multiple mutations in the S protein, designated P.1, emerged in Brazil. Here, we characterized a P.1 variant isolated in Japan by using Syrian hamsters, a well-established small animal model for the study of SARS-CoV-2 disease (COVID-19). In hamsters, the variant showed replicative abilities and pathogenicity similar to those of early and contemporary strains (i.e., SARS-CoV-2 bearing aspartic acid [D] or glycine [G] at position 614 of the S protein). Sera and/or plasma from convalescent patients and BNT162b2 messenger RNA vaccinees showed comparable neutralization titers across the P.1 variant, S-614D, and S-614G strains. In contrast, the S-614D and S-614G strains were less well recognized than the P.1 variant by serum from a P.1-infected patient. Prior infection with S-614D or S-614G strains efficiently prevented the replication of the P.1 variant in the lower respiratory tract of hamsters upon reinfection. In addition, passive transfer of neutralizing antibodies to hamsters infected with the P.1 variant or the S-614G strain led to reduced virus replication in the lower respiratory tract. However, the effect was less pronounced against the P.1 variant than the S-614G strain. These findings suggest that the P.1 variant may be somewhat antigenically different from the early and contemporary strains of SARS-CoV-2.


Asunto(s)
COVID-19/virología , SARS-CoV-2/fisiología , SARS-CoV-2/patogenicidad , Replicación Viral , Animales , Anticuerpos Neutralizantes , COVID-19/diagnóstico por imagen , COVID-19/patología , Cricetinae , Humanos , Inmunogenicidad Vacunal , Pulmón/patología , Mesocricetus , Ratones , Glicoproteína de la Espiga del Coronavirus/genética , Microtomografía por Rayos X
6.
J Antimicrob Chemother ; 78(7): 1649-1657, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37209424

RESUMEN

OBJECTIVES: Widespread resistance of influenza viruses to neuraminidase (NA) inhibitor or polymerase inhibitor, baloxavir, is a major public health concern. The amino acid mutations R152K in NA and I38T in polymerase acidic (PA) are responsible for resistance to NA inhibitors and baloxavir, respectively. METHODS: We generated recombinant A(H1N1)pdm09 viruses possessing NA-R152K, PA-I38T or both mutations by using a plasmid-based reverse genetics system, characterized their virological properties in vitro and in vivo, and examined whether oseltamivir, baloxavir and favipiravir are effective against these mutant viruses. RESULTS: The three mutant viruses showed similar or superior growth kinetics and virulence to those of wild-type virus. Although oseltamivir and baloxavir blocked the replication of the wild-type virus in vitro, oseltamivir and baloxavir failed to suppress the replication of the NA-R152K and PA-I38T viruses in vitro, respectively. Mutant virus possessing both mutations grew in the presence of oseltamivir or baloxavir in vitro. Baloxavir treatment protected mice from lethal infection with wild-type or NA-R152K virus, but failed to protect mice from lethal infection with PA-I38T or PA-I38T/NA-R152K virus. Favipiravir treatment protected mice from lethal infection with all viruses tested, whereas oseltamivir treatment did not protect at all. CONCLUSIONS: Our findings indicate that favipiravir should be used to treat patients with suspected baloxavir-resistant virus infection.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Animales , Ratones , Humanos , Oseltamivir/farmacología , Neuraminidasa/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Antivirales/farmacología , Piridonas/farmacología , Inhibidores Enzimáticos/farmacología , Triazinas/farmacología , Guanidinas/farmacología , Gripe Humana/tratamiento farmacológico , Farmacorresistencia Viral/genética
7.
J Virol ; 96(4): e0155121, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34818068

RESUMEN

Despite various attempts to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients with COVID-19 convalescent plasmas, neither appropriate approach nor clinical utility has been established. We examined the efficacy of administration of highly neutralizing COVID-19 convalescent plasma (hn-plasmas) and such plasma-derived IgG administration using the Syrian hamster COVID-19 model. Two hn-plasmas, which were in the best 1% of 340 neutralizing activity-determined convalescent plasmas, were intraperitoneally administered to SARS-CoV-2-infected hamsters, resulting in a significant reduction of viral titers in lungs by up to 32-fold compared to the viral titers in hamsters receiving control nonneutralizing plasma, while with two moderately neutralizing plasmas (mn-plasmas) administered, viral titer reduction was by up to 6-fold. IgG fractions purified from the two hn-plasmas also reduced viral titers in lungs more than those from the two mn-plasmas. The severity of lung lesions seen in hamsters receiving hn-plasmas was minimal to moderate as assessed using microcomputerized tomography, which histological examination confirmed. Western blotting revealed that all four COVID-19 convalescent plasmas variably contained antibodies against SARS-CoV-2 components, including the receptor-binding domain and S1 domain. The present data strongly suggest that administering potent neutralizing activity-confirmed COVID-19 convalescent plasmas would be efficacious in treating patients with COVID-19. IMPORTANCE Convalescent plasmas obtained from patients who recovered from a specific infection have been used as agents to treat other patients infected with the very pathogen. To treat using convalescent plasmas, despite that more than 10 randomized controlled clinical trials have been conducted and more than 100 studies are currently ongoing, the effects of convalescent plasma against COVID-19 remained uncertain. On the other hand, certain COVID-19 vaccines have been shown to reduce the clinical COVID-19 onset by 94 to 95%, for which the elicited SARS-CoV-2-neutralizing antibodies are apparently directly responsible. Here, we demonstrate that highly neutralizing effect-confirmed convalescent plasmas significantly reduce the viral titers in the lung of SARS-CoV-2-infected Syrian hamsters and block the development of virally induced lung lesions. The present data provide a proof of concept that the presence of highly neutralizing antibody in COVID-19 convalescent plasmas is directly responsible for the reduction of viral replication and support the use of highly neutralizing antibody-containing plasmas in COVID-19 therapy with convalescent plasmas.


Asunto(s)
COVID-19/terapia , Pulmón , SARS-CoV-2/fisiología , Replicación Viral , Animales , COVID-19/metabolismo , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Inmunización Pasiva , Pulmón/metabolismo , Pulmón/virología , Masculino , Mesocricetus , Células Vero , Sueroterapia para COVID-19
8.
Proc Natl Acad Sci U S A ; 117(28): 16587-16595, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32571934

RESUMEN

At the end of 2019, a novel coronavirus (severe acute respiratory syndrome coronavirus 2; SARS-CoV-2) was detected in Wuhan, China, that spread rapidly around the world, with severe consequences for human health and the global economy. Here, we assessed the replicative ability and pathogenesis of SARS-CoV-2 isolates in Syrian hamsters. SARS-CoV-2 isolates replicated efficiently in the lungs of hamsters, causing severe pathological lung lesions following intranasal infection. In addition, microcomputed tomographic imaging revealed severe lung injury that shared characteristics with SARS-CoV-2-infected human lung, including severe, bilateral, peripherally distributed, multilobular ground glass opacity, and regions of lung consolidation. SARS-CoV-2-infected hamsters mounted neutralizing antibody responses and were protected against subsequent rechallenge with SARS-CoV-2. Moreover, passive transfer of convalescent serum to naïve hamsters efficiently suppressed the replication of the virus in the lungs even when the serum was administrated 2 d postinfection of the serum-treated hamsters. Collectively, these findings demonstrate that this Syrian hamster model will be useful for understanding SARS-CoV-2 pathogenesis and testing vaccines and antiviral drugs.


Asunto(s)
Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Pulmón/patología , Neumonía Viral/virología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Betacoronavirus/patogenicidad , Betacoronavirus/fisiología , COVID-19 , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/terapia , Cricetinae , Humanos , Inmunización Pasiva , Pulmón/diagnóstico por imagen , Pulmón/virología , Mesocricetus , Pandemias , Neumonía Viral/patología , Ribonucleoproteínas/química , SARS-CoV-2 , Células Vero , Proteínas Virales/química , Replicación Viral , Sueroterapia para COVID-19
10.
J Infect Dis ; 221(10): 1699-1702, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-31837268

RESUMEN

BACKGROUND: Immunocompromised patients infected with influenza virus require prolonged treatment with neuraminidase inhibitors, because these patients are not able to eradicate the virus from the respiratory tract, leading to the emergence of drug-resistant mutant viruses. METHODS: In this study, we examined the efficacy of baloxavir marboxil in nude mice that were immunologically deficient. RESULTS: Daily treatment with a suboptimal dose of baloxavir marboxil increased the survival time of the virus-infected nude mice but did not clear the virus from their respiratory organs, resulting in gradual body weight loss after termination of treatment. CONCLUSIONS: Despite the prolonged baloxavir marboxil treatment, few resistant mutants were detected.


Asunto(s)
Antivirales/uso terapéutico , Dibenzotiepinas/uso terapéutico , Virus de la Influenza A , Morfolinas/uso terapéutico , Infecciones por Orthomyxoviridae/veterinaria , Piridonas/uso terapéutico , Triazinas/uso terapéutico , Animales , Antivirales/administración & dosificación , Dibenzotiepinas/administración & dosificación , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Desnudos , Morfolinas/administración & dosificación , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Piridonas/administración & dosificación , Triazinas/administración & dosificación
11.
J Infect Dis ; 222(7): 1155-1164, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32433769

RESUMEN

The avian influenza A(H7N9) virus has caused high mortality rates in humans, especially in the elderly; however, little is known about the mechanistic basis for this. In the current study, we used nonhuman primates to evaluate the effect of aging on the pathogenicity of A(H7N9) virus. We observed that A(H7N9) virus infection of aged animals (defined as age 20-26 years) caused more severe symptoms than infection of young animals (defined as age 2-3 years). In aged animals, lung inflammation was weak and virus infection was sustained. Although cytokine and chemokine expression in the lungs of most aged animals was lower than that in the lungs of young animals, 1 aged animal showed severe symptoms and dysregulated proinflammatory cytokine and chemokine production. These results suggest that attenuated or dysregulated immune responses in aged animals are responsible for the severe symptoms observed among elderly patients infected with A(H7N9) virus.


Asunto(s)
Envejecimiento , Subtipo H7N9 del Virus de la Influenza A , Pulmón/patología , Infecciones por Orthomyxoviridae/virología , Animales , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Pulmón/inmunología , Pulmón/virología , Macaca fascicularis , Infecciones por Orthomyxoviridae/inmunología , Replicación Viral
13.
J Gen Virol ; 100(10): 1345-1349, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31424377

RESUMEN

Long-term culture of the human lung adenocarcinoma cell line A549 promotes the differentiation of these cells toward an alveolar type II cell phenotype. Here, we evaluated the susceptibility of long-term cultured A549 cells to human influenza viruses. A549 cells were cultured continuously for 25 days (D25-A549) or 1 day (D1-A549) in Ham's F12K medium. Six human influenza A viruses grew much faster in D25-A549 cells than in D1-A549 cells; however, two influenza B viruses replicated poorly in both cell types. Two avian influenza viruses replicated efficiently in both cell types, with similar titres. Expression levels of human virus receptors were higher in D25-A549 cells than in D1-A549 cells. D25-A549 cells thus more efficiently support the replication of human influenza A viruses compared with D1-A549 cells. Our data suggest that long-term cultured A549 cells will be useful for influenza A virus research.


Asunto(s)
Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza B/crecimiento & desarrollo , Replicación Viral , Células A549 , Medios de Cultivo/química , Medios de Cultivo/metabolismo , Humanos , Virus de la Influenza A/fisiología , Virus de la Influenza B/fisiología , Gripe Humana/virología , Factores de Tiempo , Cultivo de Virus
14.
Nature ; 501(7468): 551-5, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23842494

RESUMEN

Avian influenza A viruses rarely infect humans; however, when human infection and subsequent human-to-human transmission occurs, worldwide outbreaks (pandemics) can result. The recent sporadic infections of humans in China with a previously unrecognized avian influenza A virus of the H7N9 subtype (A(H7N9)) have caused concern owing to the appreciable case fatality rate associated with these infections (more than 25%), potential instances of human-to-human transmission, and the lack of pre-existing immunity among humans to viruses of this subtype. Here we characterize two early human A(H7N9) isolates, A/Anhui/1/2013 (H7N9) and A/Shanghai/1/2013 (H7N9); hereafter referred to as Anhui/1 and Shanghai/1, respectively. In mice, Anhui/1 and Shanghai/1 were more pathogenic than a control avian H7N9 virus (A/duck/Gunma/466/2011 (H7N9); Dk/GM466) and a representative pandemic 2009 H1N1 virus (A/California/4/2009 (H1N1pdm09); CA04). Anhui/1, Shanghai/1 and Dk/GM466 replicated well in the nasal turbinates of ferrets. In nonhuman primates, Anhui/1 and Dk/GM466 replicated efficiently in the upper and lower respiratory tracts, whereas the replicative ability of conventional human influenza viruses is typically restricted to the upper respiratory tract of infected primates. By contrast, Anhui/1 did not replicate well in miniature pigs after intranasal inoculation. Critically, Anhui/1 transmitted through respiratory droplets in one of three pairs of ferrets. Glycan arrays showed that Anhui/1, Shanghai/1 and A/Hangzhou/1/2013 (H7N9) (a third human A(H7N9) virus tested in this assay) bind to human virus-type receptors, a property that may be critical for virus transmissibility in ferrets. Anhui/1 was found to be less sensitive in mice to neuraminidase inhibitors than a pandemic H1N1 2009 virus, although both viruses were equally susceptible to an experimental antiviral polymerase inhibitor. The robust replicative ability in mice, ferrets and nonhuman primates and the limited transmissibility in ferrets of Anhui/1 suggest that A(H7N9) viruses have pandemic potential.


Asunto(s)
Virus de la Influenza A , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Replicación Viral , Animales , Antivirales/farmacología , Células Cultivadas , Pollos/virología , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Perros , Inhibidores Enzimáticos/farmacología , Femenino , Hurones/virología , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/enzimología , Virus de la Influenza A/química , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/patogenicidad , Gripe Humana/tratamiento farmacológico , Macaca fascicularis/virología , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Enfermedades de los Monos/patología , Enfermedades de los Monos/virología , Neuraminidasa/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/transmisión , Codorniz/virología , Porcinos/virología , Porcinos Enanos/virología , Replicación Viral/efectos de los fármacos
15.
J Infect Dis ; 217(6): 887-896, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29186472

RESUMEN

Background: Treatment of immunocompromised, influenza virus-infected patients with the viral neuraminidase inhibitor oseltamivir often leads to the emergence of drug-resistant variants. Combination therapy with compounds that target different steps in the viral life cycle may improve treatment outcomes and reduce the emergence of drug-resistant variants. Methods: Here, we infected immunocompromised nude mice with an influenza A virus and treated them with neuraminidase (oseltamivir, laninamivir) or viral polymerase (favipiravir) inhibitors, or combinations thereof. Results: Combination therapy for 28 days increased survival times compared with monotherapy, but the animals died after treatment was terminated. Mono- and combination therapies did not consistently reduce lung virus titers. Prolonged viral replication led to the emergence of neuraminidase inhibitor-resistant variants, although viruses remained sensitive to favipiravir. Overall, favipiravir provided greater benefit than neuraminidase inhibitors. Conclusions: Collectively, our data demonstrate that combination therapy in immunocompromised hosts increases survival times, but does not suppress the emergence of neuraminidase inhibitor-resistant variants.


Asunto(s)
Amidas/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/uso terapéutico , Pirazinas/uso terapéutico , Zanamivir/análogos & derivados , Amidas/administración & dosificación , Animales , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Femenino , Guanidinas , Huésped Inmunocomprometido , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neuraminidasa/antagonistas & inhibidores , Inhibidores de la Síntesis del Ácido Nucleico/administración & dosificación , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Infecciones por Orthomyxoviridae/virología , Oseltamivir/administración & dosificación , Piranos , Pirazinas/administración & dosificación , Ácidos Siálicos , Zanamivir/administración & dosificación , Zanamivir/uso terapéutico
16.
Emerg Infect Dis ; 24(4): 746-750, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29553313

RESUMEN

To clarify the threat posed by emergence of highly pathogenic influenza A(H7N9) virus infection among humans, we characterized the viral polymerase complex. Polymerase basic 2-482R, polymerase basic 2-588V, and polymerase acidic-497R individually or additively enhanced virus polymerase activity, indicating that multiple replication-enhancing mutations in 1 isolate may contribute to virulence.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Humana/epidemiología , Gripe Humana/virología , Replicación Viral , Sustitución de Aminoácidos , Animales , Genes Virales , Humanos , Ratones , Mutación , Infecciones por Orthomyxoviridae/virología , Virulencia/genética
17.
Emerg Infect Dis ; 24(1): 75-86, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29260686

RESUMEN

During December 2016-February 2017, influenza A viruses of the H7N2 subtype infected ≈500 cats in animal shelters in New York, NY, USA, indicating virus transmission among cats. A veterinarian who treated the animals also became infected with feline influenza A(H7N2) virus and experienced respiratory symptoms. To understand the pathogenicity and transmissibility of these feline H7N2 viruses in mammals, we characterized them in vitro and in vivo. Feline H7N2 subtype viruses replicated in the respiratory organs of mice, ferrets, and cats without causing severe lesions. Direct contact transmission of feline H7N2 subtype viruses was detected in ferrets and cats; in cats, exposed animals were also infected via respiratory droplet transmission. These results suggest that the feline H7N2 subtype viruses could spread among cats and also infect humans. Outbreaks of the feline H7N2 viruses could, therefore, pose a risk to public health.


Asunto(s)
Enfermedades de los Gatos/virología , Subtipo H7N2 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/veterinaria , Animales , Enfermedades de los Gatos/epidemiología , Gatos , Femenino , Hurones , Humanos , Subtipo H7N2 del Virus de la Influenza A/clasificación , Subtipo H7N2 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana/epidemiología , Gripe Humana/transmisión , Gripe Humana/virología , Ratones Endogámicos BALB C , Ciudad de Nueva York/epidemiología , Infecciones por Orthomyxoviridae/virología , Filogenia , Cultivo de Virus
18.
J Virol ; 91(2)2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807225

RESUMEN

Pigs are considered a mixing vessel for the generation of novel pandemic influenza A viruses through reassortment because of their susceptibility to both avian and human influenza viruses. However, experiments to understand reassortment in pigs in detail have been limited because experiments with regular-sized pigs are difficult to perform. Miniature pigs have been used as an experimental animal model, but they are still large and require relatively large cages for housing. The microminipig is one of the smallest miniature pigs used for experiments. Introduced in 2010, microminipigs weigh around 10 kg at an early stage of maturity (6 to 7 months old) and are easy to handle. To evaluate the microminipig as an animal model for influenza A virus infection, we compared the receptor distribution of 10-week-old male pigs (Yorkshire Large White) and microminipigs. We found that both animals have SAα2,3Gal and SAα2,6Gal in their respiratory tracts, with similar distributions of both receptor types. We further found that the sensitivity of microminipigs to influenza A viruses was the same as that of larger miniature pigs. Our findings indicate that the microminipig could serve as a novel model animal for influenza A virus infection. IMPORTANCE: The microminipig is one of the smallest miniature pigs in the world and is used as an experimental animal model for life science research. In this study, we evaluated the microminipig as a novel animal model for influenza A virus infection. The distribution of influenza virus receptors in the respiratory tract of the microminipig was similar to that of the pig, and the sensitivity of microminipigs to influenza A viruses was the same as that of miniature pigs. Our findings suggest that microminipigs represent a novel animal model for influenza A virus infection.


Asunto(s)
Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/virología , Animales , Biomarcadores , Modelos Animales de Enfermedad , Masculino , Ácido N-Acetilneuramínico/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , Sistema Respiratorio/metabolismo , Sistema Respiratorio/virología , Porcinos , Enfermedades de los Porcinos/virología , Porcinos Enanos , Replicación Viral
19.
Nature ; 486(7403): 420-8, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22722205

RESUMEN

Highly pathogenic avian H5N1 influenza A viruses occasionally infect humans, but currently do not transmit efficiently among humans. The viral haemagglutinin (HA) protein is a known host-range determinant as it mediates virus binding to host-specific cellular receptors. Here we assess the molecular changes in HA that would allow a virus possessing subtype H5 HA to be transmissible among mammals. We identified a reassortant H5 HA/H1N1 virus-comprising H5 HA (from an H5N1 virus) with four mutations and the remaining seven gene segments from a 2009 pandemic H1N1 virus-that was capable of droplet transmission in a ferret model. The transmissible H5 reassortant virus preferentially recognized human-type receptors, replicated efficiently in ferrets, caused lung lesions and weight loss, but was not highly pathogenic and did not cause mortality. These results indicate that H5 HA can convert to an HA that supports efficient viral transmission in mammals; however, we do not know whether the four mutations in the H5 HA identified here would render a wholly avian H5N1 virus transmissible. The genetic origin of the remaining seven viral gene segments may also critically contribute to transmissibility in mammals. Nevertheless, as H5N1 viruses continue to evolve and infect humans, receptor-binding variants of H5N1 viruses with pandemic potential, including avian-human reassortant viruses as tested here, may emerge. Our findings emphasize the need to prepare for potential pandemics caused by influenza viruses possessing H5 HA, and will help individuals conducting surveillance in regions with circulating H5N1 viruses to recognize key residues that predict the pandemic potential of isolates, which will inform the development, production and distribution of effective countermeasures.


Asunto(s)
Adaptación Fisiológica/genética , Hurones/virología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Virus Reordenados/patogenicidad , Sistema Respiratorio/virología , Animales , Bioterrorismo/prevención & control , Aves/virología , Líquidos Corporales/virología , Línea Celular , Perros , Evolución Molecular , Femenino , Células HEK293 , Células HeLa , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Calor , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Gripe Aviar/transmisión , Gripe Aviar/virología , Gripe Humana/prevención & control , Gripe Humana/transmisión , Gripe Humana/virología , Epidemiología Molecular/métodos , Pandemias , Vigilancia de la Población/métodos , Estabilidad Proteica , Virus Reordenados/genética , Virus Reordenados/aislamiento & purificación , Virus Reordenados/fisiología , Receptores Virales/química , Receptores Virales/metabolismo , Sistema Respiratorio/anatomía & histología , Medidas de Seguridad , Zoonosis/transmisión , Zoonosis/virología
20.
J Infect Dis ; 216(5): 582-593, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28931216

RESUMEN

Antiviral compounds (eg, the neuraminidase inhibitor oseltamivir) are invaluable for the treatment of individuals infected with influenza A viruses of the H7N9 subtype (A[H7N9]), which have infected and killed hundreds of persons. However, oseltamivir treatment often leads to the emergence of resistant viruses in immunocompromised individuals. To better understand the emergence and properties of oseltamivir-resistant A(H7N9) viruses in immunosuppressed individuals, we infected immunosuppressed cynomolgus macaques with an A(H7N9) virus and treated them with oseltamivir. Disease severity and mortality were higher in immunosuppressed than in immunocompetent animals. Oseltamivir treatment at 2 different doses reduced A(H7N9) viral titers in infected animals, but even high-dose oseltamivir did not block viral replication sufficiently to suppress the emergence of resistant variants. Some resistant variants were not appreciably attenuated in cultured cells, but an oseltamivir-resistant A(H7N9) virus did not transmit among ferrets. These findings are useful for the control of A(H7N9) virus infections in clinical settings.


Asunto(s)
Farmacorresistencia Viral Múltiple , Huésped Inmunocomprometido , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Macaca fascicularis/virología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/uso terapéutico , Animales , Antivirales/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Subtipo H7N9 del Virus de la Influenza A/fisiología , Masculino , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/metabolismo , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA